Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nature ; 614(7947): 343-348, 2023 02.
Article in English | MEDLINE | ID: mdl-36697821

ABSTRACT

Transcriptional enhancer elements are responsible for orchestrating the temporal and spatial control over gene expression that is crucial for programming cell identity during development1-3. Here we describe a novel enhancer element that is important for regulating the expression of Prox1 in lymphatic endothelial cells. This evolutionarily conserved enhancer is bound by key lymphatic transcriptional regulators including GATA2, FOXC2, NFATC1 and PROX1. Genome editing of the enhancer to remove five nucleotides encompassing the GATA2-binding site resulted in perinatal death of homozygous mutant mice due to profound lymphatic vascular defects. Lymphatic endothelial cells in enhancer mutant mice exhibited reduced expression of genes characteristic of lymphatic endothelial cell identity and increased expression of genes characteristic of haemogenic endothelium, and acquired the capacity to generate haematopoietic cells. These data not only reveal a transcriptional enhancer element important for regulating Prox1 expression and lymphatic endothelial cell identity but also demonstrate that the lymphatic endothelium has haemogenic capacity, ordinarily repressed by Prox1.


Subject(s)
Endothelial Cells , Enhancer Elements, Genetic , Hematopoiesis , Lymphatic Vessels , Animals , Mice , Endothelial Cells/metabolism , Enhancer Elements, Genetic/genetics , Hematopoiesis/genetics , Homeodomain Proteins/metabolism , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Transcription Factors/metabolism
2.
EMBO J ; 42(11): e112590, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36912146

ABSTRACT

During development, the lymphatic vasculature forms as a second network derived chiefly from blood vessels. The transdifferentiation of embryonic venous endothelial cells (VECs) into lymphatic endothelial cells (LECs) is a key step in this process. Specification, differentiation and maintenance of LEC fate are all driven by the transcription factor Prox1, yet the downstream mechanisms remain to be elucidated. We here present a single-cell transcriptomic atlas of lymphangiogenesis in zebrafish, revealing new markers and hallmarks of LEC differentiation over four developmental stages. We further profile single-cell transcriptomic and chromatin accessibility changes in zygotic prox1a mutants that are undergoing a LEC-VEC fate shift. Using maternal and zygotic prox1a/prox1b mutants, we determine the earliest transcriptomic changes directed by Prox1 during LEC specification. This work altogether reveals new downstream targets and regulatory regions of the genome controlled by Prox1 and presents evidence that Prox1 specifies LEC fate primarily by limiting blood vascular and haematopoietic fate. This extensive single-cell resource provides new mechanistic insights into the enigmatic role of Prox1 and the control of LEC differentiation in development.


Subject(s)
Lymphatic Vessels , Zebrafish , Animals , Zebrafish/genetics , Homeodomain Proteins/genetics , Tumor Suppressor Proteins/genetics , Endothelial Cells , Cells, Cultured , Cell Differentiation , Lymphangiogenesis/genetics , Transcription Factors/genetics , Single-Cell Analysis
3.
Development ; 151(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38722096

ABSTRACT

During embryonic development, lymphatic endothelial cell (LEC) precursors are distinguished from blood endothelial cells by the expression of Prospero-related homeobox 1 (Prox1), which is essential for lymphatic vasculature formation in mouse and zebrafish. Prox1 expression initiation precedes LEC sprouting and migration, serving as the marker of specified LECs. Despite its crucial role in lymphatic development, Prox1 upstream regulation in LECs remains to be uncovered. SOX18 and COUP-TFII are thought to regulate Prox1 in mice by binding its promoter region. However, the specific regulation of Prox1 expression in LECs remains to be studied in detail. Here, we used evolutionary conservation and chromatin accessibility to identify enhancers located in the proximity of zebrafish prox1a active in developing LECs. We confirmed the functional role of the identified sequences through CRISPR/Cas9 mutagenesis of a lymphatic valve enhancer. The deletion of this region results in impaired valve morphology and function. Overall, our results reveal an intricate control of prox1a expression through a collection of enhancers. Ray-finned fish-specific distal enhancers drive pan-lymphatic expression, whereas vertebrate-conserved proximal enhancers refine expression in functionally distinct subsets of lymphatic endothelium.


Subject(s)
Endothelial Cells , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins , Lymphatic Vessels , Tumor Suppressor Proteins , Zebrafish Proteins , Zebrafish , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Zebrafish/genetics , Zebrafish/embryology , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Enhancer Elements, Genetic/genetics , Lymphatic Vessels/metabolism , Lymphatic Vessels/embryology , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Endothelial Cells/metabolism , Lymphangiogenesis/genetics , CRISPR-Cas Systems/genetics , Promoter Regions, Genetic/genetics , Mice
4.
Development ; 148(4)2021 02 19.
Article in English | MEDLINE | ID: mdl-33547133

ABSTRACT

Previous studies have shown that Vasohibin 1 (Vash1) is stimulated by VEGFs in endothelial cells and that its overexpression interferes with angiogenesis in vivo Recently, Vash1 was found to mediate tubulin detyrosination, a post-translational modification that is implicated in many cell functions, such as cell division. Here, we used the zebrafish embryo to investigate the cellular and subcellular mechanisms of Vash1 on endothelial microtubules during formation of the trunk vasculature. We show that microtubules within venous-derived secondary sprouts are strongly and selectively detyrosinated in comparison with other endothelial cells, and that this difference is lost upon vash1 knockdown. Vash1 depletion in zebrafish specifically affected secondary sprouting from the posterior cardinal vein, increasing endothelial cell divisions and cell number in the sprouts. We show that altering secondary sprout numbers and structure upon Vash1 depletion leads to defective lymphatic vessel formation and ectopic lymphatic progenitor specification in the zebrafish trunk.


Subject(s)
Cell Cycle Proteins/genetics , Embryonic Development/genetics , Lymphangiogenesis/genetics , Zebrafish/embryology , Zebrafish/genetics , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Conserved Sequence , Evolution, Molecular , Gene Expression Regulation, Developmental , Immunohistochemistry , Microtubules/metabolism , Models, Biological
5.
Development ; 147(18)2020 09 18.
Article in English | MEDLINE | ID: mdl-32839180

ABSTRACT

The lymphatic vasculature develops primarily from pre-existing veins. A pool of lymphatic endothelial cells (LECs) first sprouts from cardinal veins followed by migration and proliferation to colonise embryonic tissues. Although much is known about the molecular regulation of LEC fate and sprouting during early lymphangiogenesis, we know far less about the instructive and permissive signals that support LEC migration through the embryo. Using a forward genetic screen, we identified mbtps1 and sec23a, components of the COP-II protein secretory pathway, as essential for developmental lymphangiogenesis. In both mutants, LECs initially depart the cardinal vein but then fail in their ongoing migration. A key cargo that failed to be secreted in both mutants was a type II collagen (Col2a1). Col2a1 is normally secreted by notochord sheath cells, alongside which LECs migrate. col2a1a mutants displayed defects in the migratory behaviour of LECs and failed lymphangiogenesis. These studies thus identify Col2a1 as a key cargo secreted by notochord sheath cells and required for the migration of LECs. These findings combine with our current understanding to suggest that successive cell-to-cell and cell-matrix interactions regulate the migration of LECs through the embryonic environment during development.


Subject(s)
Cell Movement/physiology , Collagen Type II/metabolism , Embryo, Mammalian/metabolism , Endothelial Cells/metabolism , Lymphatic Vessels/metabolism , Zebrafish/metabolism , Animals , Cell Communication/physiology , Cell Proliferation/physiology , Lymphangiogenesis/physiology , Morphogenesis/physiology , Veins/metabolism
6.
Genes Dev ; 29(15): 1618-30, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26253536

ABSTRACT

The lymphatic vasculature plays roles in tissue fluid balance, immune cell trafficking, fatty acid absorption, cancer metastasis, and cardiovascular disease. Lymphatic vessels form by lymphangiogenesis, the sprouting of new lymphatics from pre-existing vessels, in both development and disease contexts. The apical signaling pathway in lymphangiogenesis is the VEGFC/VEGFR3 pathway, yet how signaling controls cellular transcriptional output remains unknown. We used a forward genetic screen in zebrafish to identify the transcription factor mafba as essential for lymphatic vessel development. We found that mafba is required for the migration of lymphatic precursors after their initial sprouting from the posterior cardinal vein. mafba expression is enriched in sprouts emerging from veins, and we show that mafba functions cell-autonomously during lymphatic vessel development. Mechanistically, Vegfc signaling increases mafba expression to control downstream transcription, and this regulatory relationship is dependent on the activity of SoxF transcription factors, which are essential for mafba expression in venous endothelium. Here we identify an indispensable Vegfc-SoxF-Mafba pathway in lymphatic development.


Subject(s)
Gene Expression Regulation, Developmental , Lymphangiogenesis/genetics , Lymphatic Vessels/embryology , MafB Transcription Factor/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor C/metabolism , Zebrafish Proteins/metabolism , Animals , Cell Movement/genetics , Embryo, Nonmammalian , MafB Transcription Factor/genetics , Mutation , Nerve Tissue Proteins/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism , Zebrafish/embryology , Zebrafish Proteins/genetics
7.
Development ; 146(2)2019 01 25.
Article in English | MEDLINE | ID: mdl-30642834

ABSTRACT

Mural cells (MCs) are essential for blood vessel stability and function; however, the mechanisms that regulate MC development remain incompletely understood, in particular those involved in MC specification. Here, we investigated the first steps of MC formation in zebrafish using transgenic reporters. Using pdgfrb and abcc9 reporters, we show that the onset of expression of abcc9, a pericyte marker in adult mice and zebrafish, occurs almost coincidentally with an increment in pdgfrb expression in peri-arterial mesenchymal cells, suggesting that these transcriptional changes mark the specification of MC lineage cells from naïve pdgfrblow mesenchymal cells. The emergence of peri-arterial pdgfrbhigh MCs required Notch signaling. We found that pdgfrb-positive cells express notch2 in addition to notch3, and although depletion of notch2 or notch3 failed to block MC emergence, embryos depleted of both notch2 and notch3 lost mesoderm- as well as neural crest-derived pdgfrbhigh MCs. Using reporters that read out Notch signaling and Notch2 receptor cleavage, we show that Notch activation in the mesenchyme precedes specification into pdgfrbhigh MCs. Taken together, these results show that Notch signaling is necessary for peri-arterial MC specification.


Subject(s)
Arteries/cytology , Arteries/embryology , Body Patterning , Mesoderm/embryology , Receptors, Notch/metabolism , Signal Transduction , Zebrafish/embryology , Animals , Biomarkers/metabolism , Endothelium, Vascular/metabolism , Mesoderm/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Time-Lapse Imaging , Transforming Growth Factor beta/metabolism
8.
Dev Dyn ; 249(10): 1201-1216, 2020 10.
Article in English | MEDLINE | ID: mdl-32525258

ABSTRACT

BACKGROUND: Lymphatic vessels play key roles in tissue fluid homeostasis, immune cell trafficking and in diverse disease settings. Lymphangiogenesis requires lymphatic endothelial cell (LEC) differentiation, proliferation, migration, and co-ordinated network formation, yet the transcriptional regulators underpinning these processes remain to be fully understood. The transcription factor MAFB was recently identified as essential for lymphangiogenesis in zebrafish and in cultured human LECs. MAFB is activated in response to VEGFC-VEGFR3 signaling and acts as a downstream effector. However, it remains unclear if the role of MAFB in lymphatic development is conserved in the mammalian embryo. RESULTS: We generated a Mafb loss-of-function mouse using CRISPR/Cas9 gene editing. Mafb mutant mice presented with perinatal lethality associated with cyanosis. We identify a role for MAFB in modifying lymphatic network morphogenesis in the developing dermis, as well as developing and postnatal diaphragm. Furthermore, mutant vessels displayed excessive smooth muscle cell coverage, suggestive of a defect in the maturation of lymphatic networks. CONCLUSIONS: This work confirms a conserved role for MAFB in murine lymphatics that is subtle and modulatory and may suggest redundancy in MAF family transcription factors during lymphangiogenesis.


Subject(s)
Lymphangiogenesis/physiology , Lymphatic Vessels/metabolism , MafB Transcription Factor/physiology , Animals , CRISPR-Cas Systems , Crosses, Genetic , Genome , Genotype , In Situ Hybridization , Mice , Mice, Knockout , Mutation , RNA, Messenger/metabolism , Signal Transduction , Time Factors
9.
Development ; 141(6): 1239-49, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24523457

ABSTRACT

The VEGFC/VEGFR3 signaling pathway is essential for lymphangiogenesis (the formation of lymphatic vessels from pre-existing vasculature) during embryonic development, tissue regeneration and tumor progression. The recently identified secreted protein CCBE1 is indispensible for lymphangiogenesis during development. The role of CCBE1 orthologs is highly conserved in zebrafish, mice and humans with mutations in CCBE1 causing generalized lymphatic dysplasia and lymphedema (Hennekam syndrome). To date, the mechanism by which CCBE1 acts remains unknown. Here, we find that ccbe1 genetically interacts with both vegfc and vegfr3 in zebrafish. In the embryo, phenotypes driven by increased Vegfc are suppressed in the absence of Ccbe1, and Vegfc-driven sprouting is enhanced by local Ccbe1 overexpression. Moreover, Vegfc- and Vegfr3-dependent Erk signaling is impaired in the absence of Ccbe1. Finally, CCBE1 is capable of upregulating the levels of fully processed, mature VEGFC in vitro and the overexpression of mature VEGFC rescues ccbe1 loss-of-function phenotypes in zebrafish. Taken together, these data identify Ccbe1 as a crucial component of the Vegfc/Vegfr3 pathway in the embryo.


Subject(s)
Lymphangiogenesis/physiology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , DNA/genetics , Gene Expression Regulation, Developmental , Humans , Lymphangiogenesis/genetics , MAP Kinase Signaling System , Mice , Molecular Sequence Data , Point Mutation , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Signal Transduction , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
10.
Hum Mol Genet ; 23(5): 1286-97, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24163130

ABSTRACT

Mutations in SOX18, VEGFC and Vascular Endothelial Growth Factor 3 underlie the hereditary lymphatic disorders hypotrichosis-lymphedema-telangiectasia (HLT), Milroy-like lymphedema and Milroy disease, respectively. Genes responsible for hereditary lymphedema are key regulators of lymphatic vascular development in the embryo. To identify novel modulators of lymphangiogenesis, we used a mouse model of HLT (Ragged Opossum) and performed gene expression profiling of aberrant dermal lymphatic vessels. Expression studies and functional analysis in zebrafish and mice revealed one candidate, ArfGAP with RhoGAP domain, Ankyrin repeat and PH domain 3 (ARAP3), which is down-regulated in HLT mouse lymphatic vessels and necessary for lymphatic vascular development in mice and zebrafish. We position this known regulator of cell behaviour during migration as a mediator of the cellular response to Vegfc signalling in lymphatic endothelial cells in vitro and in vivo. Our data refine common mechanisms that are likely to contribute during both development and the pathogenesis of lymphatic vascular disorders.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , GTPase-Activating Proteins/genetics , Gene Expression Regulation , Hypotrichosis/genetics , Lymphangiogenesis/genetics , Lymphedema/genetics , Telangiectasis/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Movement/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Female , GTPase-Activating Proteins/metabolism , Lymphatic Vessels/metabolism , Mice , Mice, Knockout , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Syndrome , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Zebrafish
11.
Development ; 140(9): 1857-70, 2013 May.
Article in English | MEDLINE | ID: mdl-23571211

ABSTRACT

The lymphatic vascular system develops from the pre-existing blood vasculature of the vertebrate embryo. New insights into lymphatic vascular development have recently been achieved with the use of alternative model systems, new molecular tools, novel imaging technologies and growing interest in the role of lymphatic vessels in human disorders. The signals and cellular mechanisms that facilitate the emergence of lymphatic endothelial cells from veins, guide migration through the embryonic environment, mediate interactions with neighbouring tissues and control vessel maturation are beginning to emerge. Here, we review the most recent advances in lymphatic vascular development, with a major focus on mouse and zebrafish model systems.


Subject(s)
Lymphangiogenesis , Lymphatic Vessels/embryology , Morphogenesis , Animals , Biological Evolution , Cell Movement , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Ephrin-B2/genetics , Ephrin-B2/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lymphatic Vessels/cytology , Mice , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
12.
Development ; 140(9): 1912-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23515471

ABSTRACT

Tightly controlled DNA replication and RNA transcription are essential for differentiation and tissue growth in multicellular organisms. Histone chaperones, including the FACT (facilitates chromatin transcription) complex, are central for these processes and act by mediating DNA access through nucleosome reorganisation. However, their roles in vertebrate organogenesis are poorly understood. Here, we report the identification of zebrafish mutants for the gene encoding Structure specific recognition protein 1a (Ssrp1a), which, together with Spt16, forms the FACT heterodimer. Focussing on the liver and eye, we show that zygotic Ssrp1a is essential for proliferation and differentiation during organogenesis. Specifically, gene expression indicative of progressive organ differentiation is disrupted and RNA transcription is globally reduced. Ssrp1a-deficient embryos exhibit DNA synthesis defects and prolonged S phase, uncovering a role distinct from that of Spt16, which promotes G1 phase progression. Gene deletion/replacement experiments in Drosophila show that Ssrp1b, Ssrp1a and N-terminal Ssrp1a, equivalent to the yeast homologue Pob3, can substitute Drosophila Ssrp function. These data suggest that (1) Ssrp1b does not compensate for Ssrp1a loss in the zebrafish embryo, probably owing to insufficient expression levels, and (2) despite fundamental structural differences, the mechanisms mediating DNA accessibility by FACT are conserved between yeast and metazoans. We propose that the essential functions of Ssrp1a in DNA replication and gene transcription, together with its dynamic spatiotemporal expression, ensure organ-specific differentiation and proportional growth, which are crucial for the forming embryo.


Subject(s)
Cell Cycle , Organogenesis , Transcription, Genetic , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Proliferation , Chromatin Assembly and Disassembly , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila/embryology , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Endoderm/cytology , Endoderm/embryology , Endoderm/metabolism , Eye/cytology , Eye/embryology , Eye/metabolism , Female , Gene Expression Regulation, Developmental , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Imaginal Discs/cytology , Imaginal Discs/embryology , Imaginal Discs/metabolism , Liver/cytology , Liver/embryology , Liver/metabolism , Male , Mitotic Index , Mutation , RNA/biosynthesis , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
13.
Blood ; 123(7): 1102-12, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24269955

ABSTRACT

Vascular endothelial growth factor-D (VEGFD) is a potent pro-lymphangiogenic molecule during tumor growth and is considered a key therapeutic target to modulate metastasis. Despite roles in pathological neo-lymphangiogenesis, the characterization of an endogenous role for VEGFD in vascular development has remained elusive. Here, we used zebrafish to assay for genetic interactions between the Vegf/Vegf-receptor pathway and SoxF transcription factors and identified a specific interaction between Vegfd and Sox18. Double knockdown zebrafish embryos for Sox18/Vegfd and Sox7/Vegfd exhibit defects in arteriovenous differentiation. Supporting this observation, we found that Sox18/Vegfd double but not single knockout mice displayed dramatic vascular development defects. We find that VEGFD-mitogen-activated protein kinase kinase-extracellular signal-regulated kinase signaling modulates SOX18-mediated transcription, functioning at least in part by enhancing nuclear concentration and transcriptional activity in vascular endothelial cells. This work suggests that VEGFD-mediated pathologies include or involve an underlying dysregulation of SOXF-mediated transcriptional networks.


Subject(s)
Blood Vessels/embryology , Neovascularization, Physiologic/genetics , SOXF Transcription Factors/metabolism , Vascular Endothelial Growth Factor D/physiology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Animals, Genetically Modified , Embryo, Mammalian , Embryo, Nonmammalian , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks/genetics , Male , Mice , Mice, Inbred C57BL , SOXF Transcription Factors/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
14.
Article in English | MEDLINE | ID: mdl-38940420

ABSTRACT

New technologies have resulted in a better understanding of blood and lymphatic vascular heterogeneity at the cellular and molecular levels. However, we still need to learn more about the heterogeneity of the cardiovascular and lymphatic systems among different species at the anatomical and functional levels. Even the deceptively simple question of the functions of fish lymphatic vessels has yet to be conclusively answered. The most common interpretation assumes a similar dual setup of the vasculature in zebrafish and mammals: a cardiovascular circulatory system, and a lymphatic vascular system (LVS), in which the unidirectional flow is derived from surplus interstitial fluid and returned into the cardiovascular system. A competing interpretation questions the identity of the lymphatic vessels in fish as at least some of them receive their flow from arteries via specialised anastomoses, neither requiring an interstitial source for the lymphatic flow nor stipulating unidirectionality. In this alternative view, the 'fish lymphatics' are a specialised subcompartment of the cardiovascular system, called the secondary vascular system (SVS). Many of the contradictions found in the literature appear to stem from the fact that the SVS develops in part or completely from an embryonic LVS by transdifferentiation. Future research needs to establish the extent of embryonic transdifferentiation of lymphatics into SVS blood vessels. Similarly, more insight is needed into the molecular regulation of vascular development in fish. Most fish possess more than the five vascular endothelial growth factor (VEGF) genes and three VEGF receptor genes that we know from mice or humans, and the relative tolerance of fish to whole-genome and gene duplications could underlie the evolutionary diversification of the vasculature. This review discusses the key elements of the fish lymphatics versus the SVS and attempts to draw a picture coherent with the existing data, including phylogenetic knowledge.

15.
Front Cell Dev Biol ; 10: 891538, 2022.
Article in English | MEDLINE | ID: mdl-35615697

ABSTRACT

Epigenetic regulation is integral in orchestrating the spatiotemporal regulation of gene expression which underlies tissue development. The emergence of new tools to assess genome-wide epigenetic modifications has enabled significant advances in the field of vascular biology in zebrafish. Zebrafish represents a powerful model to investigate the activity of cis-regulatory elements in vivo by combining technologies such as ATAC-seq, ChIP-seq and CUT&Tag with the generation of transgenic lines and live imaging to validate the activity of these regulatory elements. Recently, this approach led to the identification and characterization of key enhancers of important vascular genes, such as gata2a, notch1b and dll4. In this review we will discuss how the latest technologies in epigenetics are being used in the zebrafish to determine chromatin states and assess the function of the cis-regulatory sequences that shape the zebrafish vascular network.

16.
Neuro Oncol ; 24(5): 726-738, 2022 05 04.
Article in English | MEDLINE | ID: mdl-34919147

ABSTRACT

BACKGROUND: Patient-derived xenograft (PDX) models of glioblastoma (GBM) are a central tool for neuro-oncology research and drug development, enabling the detection of patient-specific differences in growth, and in vivo drug response. However, existing PDX models are not well suited for large-scale or automated studies. Thus, here, we investigate if a fast zebrafish-based PDX model, supported by longitudinal, AI-driven image analysis, can recapitulate key aspects of glioblastoma growth and enable case-comparative drug testing. METHODS: We engrafted 11 GFP-tagged patient-derived GBM IDH wild-type cell cultures (PDCs) into 1-day-old zebrafish embryos, and monitored fish with 96-well live microscopy and convolutional neural network analysis. Using light-sheet imaging of whole embryos, we analyzed further the invasive growth of tumor cells. RESULTS: Our pipeline enables automatic and robust longitudinal observation of tumor growth and survival of individual fish. The 11 PDCs expressed growth, invasion and survival heterogeneity, and tumor initiation correlated strongly with matched mouse PDX counterparts (Spearman R = 0.89, p < 0.001). Three PDCs showed a high degree of association between grafted tumor cells and host blood vessels, suggesting a perivascular invasion phenotype. In vivo evaluation of the drug marizomib, currently in clinical trials for GBM, showed an effect on fish survival corresponding to PDC in vitro and in vivo marizomib sensitivity. CONCLUSIONS: Zebrafish xenografts of GBM, monitored by AI methods in an automated process, present a scalable alternative to mouse xenograft models for the study of glioblastoma tumor initiation, growth, and invasion, applicable to patient-specific drug evaluation.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Glioblastoma/pathology , Heterografts , Humans , Xenograft Model Antitumor Assays , Zebrafish
17.
Cell Rep ; 39(12): 110982, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35732122

ABSTRACT

Lymphangiogenesis, formation of lymphatic vessels from pre-existing vessels, is a dynamic process that requires cell migration. Regardless of location, migrating lymphatic endothelial cell (LEC) progenitors probe their surroundings to form the lymphatic network. Lymphatic-development regulation requires the transcription factor MAFB in different species. Zebrafish Mafba, expressed in LEC progenitors, is essential for their migration in the trunk. However, the transcriptional mechanism that orchestrates LEC migration in different lymphatic endothelial beds remains elusive. Here, we uncover topographically different requirements of the two paralogs, Mafba and Mafbb, for LEC migration. Both mafba and mafbb are necessary for facial lymphatic development, but mafbb is dispensable for trunk lymphatic development. On the molecular level, we demonstrate a regulatory network where Vegfc-Vegfd-SoxF-Mafba-Mafbb is essential in facial lymphangiogenesis. We identify that mafba and mafbb tune the directionality of LEC migration and vessel morphogenesis that is ultimately necessary for lymphatic function.


Subject(s)
Lymphatic Vessels , Zebrafish , Animals , Cell Movement , Endothelial Cells , Lymphangiogenesis , Morphogenesis , Signal Transduction
18.
Elife ; 112022 07 21.
Article in English | MEDLINE | ID: mdl-35861713

ABSTRACT

Dysfunctional and leaky blood vessels resulting from disruption of the endothelial cell (EC) barrier accompanies numerous diseases. The EC barrier is established through endothelial cell tight and adherens junctions. However, the expression pattern and precise contribution of different junctional proteins to the EC barrier is poorly understood. Here, we focus on organs with continuous endothelium to identify structural and functional in vivo characteristics of the EC barrier. Assembly of multiple single-cell RNAseq datasets into a single integrated database revealed the variability and commonalities of EC barrier patterning. Across tissues, Claudin5 exhibited diminishing expression along the arteriovenous axis, correlating with EC barrier integrity. Functional analysis identified tissue-specific differences in leakage properties and response to the leakage agonist histamine. Loss of Claudin5 enhanced histamine-induced leakage in an organotypic and vessel type-specific manner in an inducible, EC-specific, knock-out mouse. Mechanistically, Claudin5 loss left junction ultrastructure unaffected but altered its composition, with concomitant loss of zonula occludens-1 and upregulation of VE-Cadherin expression. These findings uncover the organ-specific organisation of the EC barrier and distinct importance of Claudin5 in different vascular beds, providing insights to modify EC barrier stability in a targeted, organ-specific manner.


Subject(s)
Adherens Junctions , Claudin-5/metabolism , Histamine , Adherens Junctions/metabolism , Animals , Cadherins/metabolism , Endothelial Cells/metabolism , Endothelium/metabolism , Histamine/metabolism , Mice , Tight Junctions/metabolism
19.
Elife ; 112022 03 22.
Article in English | MEDLINE | ID: mdl-35316177

ABSTRACT

The migration of lymphatic endothelial cells (LECs) is key for the development of the complex and vast lymphatic vascular network that pervades most tissues in an organism. In zebrafish, arterial intersegmental vessels together with chemokines have been shown to promote lymphatic cell migration from the horizontal myoseptum (HM). We observed that emergence of mural cells around the intersegmental arteries coincides with lymphatic departure from HM which raised the possibility that arterial mural cells promote LEC migration. Our live imaging and cell ablation experiments revealed that LECs migrate slower and fail to establish the lymphatic vascular network in the absence of arterial mural cells. We determined that mural cells are a source for the C-X-C motif chemokine 12 (Cxcl12a and Cxcl12b), vascular endothelial growth factor C (Vegfc) and collagen and calcium-binding EGF domain-containing protein 1 (Ccbe1). We showed that chemokine and growth factor signalling function cooperatively to induce robust LEC migration. Specifically, Vegfc-Vegfr3 signalling, but not chemokines, induces extracellular signal-regulated kinase (ERK) activation in LECs, and has an additional pro-survival role in LECs during the migration. Together, the identification of mural cells as a source for signals that guide LEC migration and survival will be important in the future design for rebuilding lymphatic vessels in disease contexts.


Subject(s)
Endothelial Cells , Vascular Endothelial Growth Factor C , Animals , Arteries , Cues , Endothelial Cells/physiology , Vascular Endothelial Growth Factor C/physiology , Zebrafish
20.
Trends Mol Med ; 27(10): 955-970, 2021 10.
Article in English | MEDLINE | ID: mdl-34332911

ABSTRACT

The lymphatic vasculature is emerging as a multifaceted regulator of tissue homeostasis and regeneration. Lymphatic vessels drain fluid, macromolecules, and immune cells from peripheral tissues to lymph nodes (LNs) and the systemic circulation. Their recently uncovered functions extend beyond drainage and include direct modulation of adaptive immunity and paracrine regulation of organ growth. The developmental mechanisms controlling lymphatic vessel growth have been described with increasing precision. It is less clear how the essential functional features of lymphatic vessels are established and maintained. We discuss the mechanisms that maintain lymphatic vessel integrity in adult tissues and control vessel repair and regeneration. This knowledge is crucial for understanding the pathological vessel changes that contribute to disease, and provides an opportunity for therapy development.


Subject(s)
Lymphangiogenesis , Lymphatic Vessels , Homeostasis , Humans , Lymphangiogenesis/physiology , Lymphatic Vessels/physiology
SELECTION OF CITATIONS
SEARCH DETAIL